1
|
McGill JL, Loving CL, Kehrli ME. Future of Immune Modulation in Animal Agriculture. Annu Rev Anim Biosci 2025; 13:255-275. [PMID: 39159206 DOI: 10.1146/annurev-animal-111523-102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Immune modulation in animal agriculture has been of research interest for several decades, yet only a few immunomodulators have received regulatory approval in the United States and around the world. In this review, we summarize market and regulatory environments impacting commercial development of immunomodulators for use in livestock and poultry. In the United States, very few immunomodulators have received regulatory approval for use in livestock by either the US Department of Agriculture Center for Veterinary Biologics or the Food and Drug Administration (FDA). To date, only one immunomodulator has received FDA approval, and an extensive body of peer-reviewed literature is available regarding the basis for its use and health benefits. We present a more thorough review of the history and impact of this immune restorative. Finally, we discuss the interaction of immunomodulators on health, metabolism, and other factors impacting the future of immune modulation in livestock.
Collapse
Affiliation(s)
- Jodi L McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa, USA
| | - Crystal L Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, USDA, Ames, Iowa, USA
| | - Marcus E Kehrli
- National Animal Disease Center, Agricultural Research Service, USDA, Ames, Iowa, USA;
| |
Collapse
|
2
|
Sey EA, Warris A. The gut-lung axis: the impact of the gut mycobiome on pulmonary diseases and infections. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae008. [PMID: 39193472 PMCID: PMC11316619 DOI: 10.1093/oxfimm/iqae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 08/29/2024] Open
Abstract
The gastrointestinal tract contains a diverse microbiome consisting of bacteria, fungi, viruses and archaea. Although these microbes usually reside as commensal organisms, it is now well established that higher abundance of specific bacterial or fungal species, or loss of diversity in the microbiome can significantly affect development, progression and outcomes in disease. Studies have mainly focused on the effects of bacteria, however, the impact of other microbes, such as fungi, has received increased attention in the last few years. Fungi only represent around 0.1% of the total gut microbial population. However, key fungal taxa such as Candida, Aspergillus and Wallemia have been shown to significantly impact health and disease. The composition of the gut mycobiome has been shown to affect immunity at distal sites, such as the heart, lung, brain, pancreas, and liver. In the case of the lung this phenomenon is referred to as the 'gut-lung axis'. Recent studies have begun to explore and unveil the relationship between gut fungi and lung immunity in diseases such as asthma and lung cancer, and lung infections caused by viruses, bacteria and fungi. In this review we will summarize the current, rapidly growing, literature describing the impact of the gut mycobiome on respiratory disease and infection.
Collapse
Affiliation(s)
- Emily A Sey
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, EX4 4QD, UK
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, EX4 4QD, UK
| |
Collapse
|
3
|
Yang H, Fan X, Mao X, Yu B, He J, Yan H, Wang J. The protective role of prebiotics and probiotics on diarrhea and gut damage in the rotavirus-infected piglets. J Anim Sci Biotechnol 2024; 15:61. [PMID: 38698473 PMCID: PMC11067158 DOI: 10.1186/s40104-024-01018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/29/2024] [Indexed: 05/05/2024] Open
Abstract
Rotavirus is one of the pathogenic causes that induce diarrhea in young animals, especially piglets, worldwide. However, nowadays, there is no specific drug available to treat the disease, and the related vaccines have no obvious efficiency in some countries. Via analyzing the pathogenesis of rotavirus, it inducing diarrhea is mainly due to disturb enteric nervous system, destroy gut mucosal integrity, induce intracellular electrolyte imbalance, and impair gut microbiota and immunity. Many studies have already proved that prebiotics and probiotics can mitigate the damage and diarrhea induced by rotavirus infection in hosts. Based on these, the current review summarizes and discusses the effects and mechanisms of prebiotics and probiotics on rotavirus-induced diarrhea in piglets. This information will highlight the basis for the swine production utilization of prebiotics and probiotics in the prevention or treatment of rotavirus infection in the future.
Collapse
Affiliation(s)
- Heng Yang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Xiangqi Fan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China.
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| |
Collapse
|
4
|
Zhu X, Zhang C, Feng S, He R, Zhang S. Intestinal microbiota regulates the gut-thyroid axis: the new dawn of improving Hashimoto thyroiditis. Clin Exp Med 2024; 24:39. [PMID: 38386169 PMCID: PMC10884059 DOI: 10.1007/s10238-024-01304-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Intestinal microbiota plays an indispensable role in the host's innate immune system, which may be related to the occurrence of many autoimmune diseases. Hashimoto thyroiditis (HT) is one of the most common autoimmune diseases, and there is plenty of evidence indicating that HT may be related to genetics and environmental triggers, but the specific mechanism has not been proven clearly. Significantly, the composition and abundance of intestinal microbiota in patients with HT have an obvious difference. This phenomenon led us to think about whether intestinal microbiota can affect the progress of HT through some mechanisms. By summarizing the potential mechanism of intestinal microflora in regulating Hashimoto thyroiditis, this article explores the possibility of improving HT by regulating intestinal microbiota and summarizes relevant biomarkers as therapeutic targets, which provide new ideas for the clinical diagnosis and treatment of Hashimoto thyroiditis.
Collapse
Affiliation(s)
- Xiaxin Zhu
- Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Chi Zhang
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310018, People's Republic of China
| | - Shuyan Feng
- Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Ruonan He
- Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Shuo Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University (The Xin Hua Hospital of Zhejiang Province), No. 318 Chaowang Road, Hangzhou, 310005, Zhejiang, People's Republic of China.
| |
Collapse
|
5
|
Parreno V, Bai M, Liu F, Jing J, Olney E, Li G, Wen K, Yang X, Castellucc TB, Kocher JF, Zhou X, Yuan L. Probiotic as Adjuvant Significantly Improves Protection of the Lanzhou Trivalent Rotavirus Vaccine against Heterologous Challenge in a Gnotobiotic Pig Model of Human Rotavirus Infection and Disease. Vaccines (Basel) 2022; 10:vaccines10091529. [PMID: 36146607 PMCID: PMC9506166 DOI: 10.3390/vaccines10091529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/10/2022] [Accepted: 09/11/2022] [Indexed: 11/25/2022] Open
Abstract
This preclinical study in the gnotobiotic (Gn) pig model of human rotavirus (HRV) infection and disease evaluates the effect of probiotic Lactobacillus rhamnosus GG (LGG) as a mucosal adjuvant on the immunogenicity and cross-protective efficacy of the Lanzhou live oral trivalent (G2, G3, G4) vaccine (TLV, aka LLR3). Gn pigs were immunized with three doses of TLV with or without concurrent administration of nine doses of LGG around the time of the first dose of the TLV vaccination, and were challenged orally with the virulent heterotypic Wa G1P[8] HRV. Three doses of TLV were highly immunogenic and conferred partial protection against the heterotypic HRV infection. LGG significantly enhanced the intestinal and systemic immune responses and improved the effectiveness of protection against the heterotypic HRV challenge-induced diarrhea and virus shedding. In conclusion, we demonstrated the immune-stimulating effects of probiotic LGG as a vaccine adjuvant and generated detailed knowledge regarding the cross-reactive and type-specific antibody and effector B and T cell immune responses induced by the TLV. Due to the low cost, ease of distribution and administration, and favorable safety profiles, LGG as an adjuvant has the potential to play a critical role in improving rotavirus vaccine efficacy and making the vaccines more cost-effective.
Collapse
Affiliation(s)
- Viviana Parreno
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- INCUINTA, Institutot de Virología e Innovaciones Tecnológicas (IVIT), Instituto Nacional de Tecnología Agropecuaria (INTA)-CONICET, Ciudad Autónoma de Buenos Aires C1033AAE, Argentina
| | - Muqun Bai
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Lanzhou Institute of Biological Products, Lanzhou 730046, China
| | - Fangning Liu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Jiqiang Jing
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Department of Animal Medicine, Shandong Vocational College of Animal Husbandry and Veterinary Medicine, Weifang 261071, China
| | - Erika Olney
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Guohua Li
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Ke Wen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Xingdong Yang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Tammy Bui Castellucc
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Jacob F. Kocher
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Xu Zhou
- Lanzhou Institute of Biological Products, Lanzhou 730046, China
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Correspondence: ; Tel.: +86-540-231-9053
| |
Collapse
|
6
|
Zhang H, Zhao H, Zhao Y, Sui L, Li F, Zhang H, Li J, Jiang Y, Cui W, Ding G, Zhou H, Wang L, Qiao X, Tang L, Wang X, Li Y. Auxotrophic Lactobacillus Expressing Porcine Rotavirus VP4 Constructed Using CRISPR-Cas9D10A System Induces Effective Immunity in Mice. Vaccines (Basel) 2022; 10:vaccines10091510. [PMID: 36146587 PMCID: PMC9504633 DOI: 10.3390/vaccines10091510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Porcine rotavirus (PoRV) mainly causes acute diarrhea in piglets under eight weeks of age and has potentially high morbidity and mortality rates. As vaccine carriers for oral immunization, lactic acid bacteria (LAB) are an ideal strategy for blocking PoRV infections. However, the difficulty in knocking out specific genes, inserting foreign genes, and the residues of antibiotic selection markers are major challenges for the oral vaccination of LAB. In this study, the target gene, alanine racemase (alr), in the genome of Lactobacillus casei strain W56 (L. casei W56) was knocked out to construct an auxotrophic L. casei strain (L. casei Δalr W56) using the CRISPR-Cas9D10A gene editing system. A recombinant strain (pPG-alr-VP4/Δalr W56) was constructed using an electrotransformed complementary plasmid. Expression of the alr-VP4 fusion protein from pPG-alr-VP4/Δalr W56 was detected using Western blotting. Mice orally immunized with pPG-alr-VP4/Δalr W56 exhibited high levels of serum IgG and mucosal secretory immunoglobulin A (SIgA), which exhibited neutralizing effects against PoRV. Cytokines levels in serum detected using ELISA, indicated that the recombinant strain induced an immune response dominated by Th2 cells. Our data suggest that pPG-alr-VP4/Δalr W56, an antibiotic-resistance-free LAB, provides a safer vaccine strategy against PoRV infection.
Collapse
Affiliation(s)
- Hailin Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Haiyuan Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Jiangsu Hanswine Food Co., Ltd., Ma’anshan 243000, China
| | - Yuliang Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ling Sui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Fengsai Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Huijun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jiaxuan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Wen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Guojie Ding
- Harbin Vikeses Biological Technology Co., Ltd., Harbin 150030, China
| | - Han Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Li Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Xinyuan Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Xiaona Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
- Correspondence: (X.W.); (Y.L.); Tel./Fax: +86-451-5519-0363 (Y.L.)
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
- Correspondence: (X.W.); (Y.L.); Tel./Fax: +86-451-5519-0363 (Y.L.)
| |
Collapse
|
7
|
The Combined Escherichia coli Nissle 1917 and Tryptophan Treatment Modulates Immune and Metabolome Responses to Human Rotavirus Infection in a Human Infant Fecal Microbiota-Transplanted Malnourished Gnotobiotic Pig Model. mSphere 2022; 7:e0027022. [PMID: 36073800 PMCID: PMC9599269 DOI: 10.1128/msphere.00270-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human rotavirus (HRV) is a major cause of childhood diarrhea in developing countries where widespread malnutrition contributes to the decreased oral vaccine efficacy and increased prevalence of other enteric infections, which are major concerns for global health. Neonatal gnotobiotic (Gn) piglets closely resemble human infants in their anatomy, physiology, and outbred status, providing a unique model to investigate malnutrition, supplementations, and HRV infection. To understand the molecular signatures associated with immune enhancement and reduced diarrheal severity by Escherichia coli Nissle 1917 (EcN) and tryptophan (TRP), immunological responses and global nontargeted metabolomics and lipidomics approaches were investigated on the plasma and fecal contents of malnourished pigs transplanted with human infant fecal microbiota and infected with virulent (Vir) HRV. Overall, EcN + TRP combined (rather than individual supplement action) promoted greater and balanced immunoregulatory/immunostimulatory responses associated with greater protection against HRV infection and disease in malnourished humanized piglets. Moreover, EcN + TRP treatment upregulated the production of several metabolites with immunoregulatory/immunostimulatory properties: amino acids (N-acetylserotonin, methylacetoacetyl-CoA), lipids (gamma-butyrobetaine, eicosanoids, cholesterol-sulfate, sphinganine/phytosphingosine, leukotriene), organic compound (biliverdin), benzenoids (gentisic acid, aminobenzoic acid), and nucleotides (hypoxathine/inosine/xanthine, cytidine-5'-monophosphate). Additionally, the levels of several proinflammatory metabolites of organic compounds (adenosylhomocysteine, phenylacetylglycine, urobilinogen/coproporphyrinogen) and amino acid (phenylalanine) were reduced following EcN + TRP treatment. These results suggest that the EcN + TRP effects on reducing HRV diarrhea in neonatal Gn pigs were at least in part due to altered metabolites, those involved in lipid, amino acid, benzenoids, organic compounds, and nucleotide metabolism. Identification of these important mechanisms of EcN/TRP prevention of HRV diarrhea provides novel targets for therapeutics development. IMPORTANCE Human rotavirus (HRV) is the most common cause of viral gastroenteritis in children, especially in developing countries, where the efficacy of oral HRV vaccines is reduced. Escherichia coli Nissle 1917 (EcN) is used to treat enteric infections and ulcerative colitis while tryptophan (TRP) is a biomarker of malnutrition, and its supplementation can alleviate intestinal inflammation and normalize intestinal microbiota in malnourished hosts. Supplementation of EcN + TRP to malnourished humanized gnotobiotic piglets enhanced immune responses and resulted in greater protection against HRV infection and diarrhea. Moreover, EcN + TRP supplementation increased the levels of immunoregulatory/immunostimulatory metabolites while decreasing the production of proinflammatory metabolites in plasma and fecal samples. Profiling of immunoregulatory and proinflammatory biomarkers associated with HRV perturbations will aid in the identification of treatments against HRV and other enteric diseases in malnourished children.
Collapse
|
8
|
Sadrekarimi H, Gardanova ZR, Bakhshesh M, Ebrahimzadeh F, Yaseri AF, Thangavelu L, Hasanpoor Z, Zadeh FA, Kahrizi MS. Emerging role of human microbiome in cancer development and response to therapy: special focus on intestinal microflora. Lab Invest 2022; 20:301. [PMID: 35794566 PMCID: PMC9258144 DOI: 10.1186/s12967-022-03492-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022]
Abstract
In recent years, there has been a greater emphasis on the impact of microbial populations inhabiting the gastrointestinal tract on human health and disease. According to the involvement of microbiota in modulating physiological processes (such as immune system development, vitamins synthesis, pathogen displacement, and nutrient uptake), any alteration in its composition and diversity (i.e., dysbiosis) has been linked to a variety of pathologies, including cancer. In this bidirectional relationship, colonization with various bacterial species is correlated with a reduced or elevated risk of certain cancers. Notably, the gut microflora could potentially play a direct or indirect role in tumor initiation and progression by inducing chronic inflammation and producing toxins and metabolites. Therefore, identifying the bacterial species involved and their mechanism of action could be beneficial in preventing the onset of tumors or controlling their advancement. Likewise, the microbial community affects anti-cancer approaches’ therapeutic potential and adverse effects (such as immunotherapy and chemotherapy). Hence, their efficiency should be evaluated in the context of the microbiome, underlining the importance of personalized medicine. In this review, we summarized the evidence revealing the microbiota's involvement in cancer and its mechanism. We also delineated how microbiota could predict colon carcinoma development or response to current treatments to improve clinical outcomes.
Collapse
|
9
|
Michael H, Amimo JO, Rajashekara G, Saif LJ, Vlasova AN. Mechanisms of Kwashiorkor-Associated Immune Suppression: Insights From Human, Mouse, and Pig Studies. Front Immunol 2022; 13:826268. [PMID: 35585989 PMCID: PMC9108366 DOI: 10.3389/fimmu.2022.826268] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/31/2022] [Indexed: 12/11/2022] Open
Abstract
Malnutrition refers to inadequate energy and/or nutrient intake. Malnutrition exhibits a bidirectional relationship with infections whereby malnutrition increases risk of infections that further aggravates malnutrition. Severe malnutrition (SM) is the main cause of secondary immune deficiency and mortality among children in developing countries. SM can manifest as marasmus (non-edematous), observed most often (68.6% of all malnutrition cases), kwashiorkor (edematous), detected in 23.8% of cases, and marasmic kwashiorkor, identified in ~7.6% of SM cases. Marasmus and kwashiorkor occur due to calorie-energy and protein-calorie deficiency (PCD), respectively. Kwashiorkor and marasmic kwashiorkor present with reduced protein levels, protein catabolism rates, and altered levels of micronutrients leading to uncontrolled oxidative stress, exhaustion of anaerobic commensals, and proliferation of pathobionts. Due to these alterations, kwashiorkor children present with profoundly impaired immune function, compromised intestinal barrier, and secondary micronutrient deficiencies. Kwashiorkor-induced alterations contribute to growth stunting and reduced efficacy of oral vaccines. SM is treated with antibiotics and ready-to-use therapeutic foods with variable efficacy. Kwashiorkor has been extensively investigated in gnotobiotic (Gn) mice and piglet models to understand its multiple immediate and long-term effects on children health. Due to numerous physiological and immunological similarities between pigs and humans, pig represents a highly relevant model to study kwashiorkor pathophysiology and immunology. Here we summarize the impact of kwashiorkor on children's health, immunity, and gut functions and review the relevant findings from human and animal studies. We also discuss the reciprocal interactions between PCD and rotavirus-a highly prevalent enteric childhood pathogen due to which pathogenesis and immunity are affected by childhood SM.
Collapse
Affiliation(s)
- Husheem Michael
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Joshua O. Amimo
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
- Department of Animal Production, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Gireesh Rajashekara
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Linda J. Saif
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| |
Collapse
|
10
|
Exploring the Gut Microbiome in Myasthenia Gravis. Nutrients 2022; 14:nu14081647. [PMID: 35458209 PMCID: PMC9027283 DOI: 10.3390/nu14081647] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/13/2022] Open
Abstract
The human gut microbiota is vital for maintaining human health in terms of immune system homeostasis. Perturbations in the composition and function of microbiota have been associated with several autoimmune disorders, including myasthenia gravis (MG), a neuromuscular condition associated with varying weakness and rapid fatigue of the skeletal muscles triggered by the host’s antibodies against the acetylcholine receptor (AChR) in the postsynaptic muscle membrane at the neuromuscular junction (NMJ). It is hypothesized that perturbation of the gut microbiota is associated with the pathogenesis of MG. The gut microbiota community profiles are usually generated using 16S rRNA gene sequencing. Compared to healthy individuals, MG participants had an altered gut microbiota’s relative abundance of bacterial taxa, particularly with a drop in Clostridium. The microbial diversity related to MG severity and the overall fecal short-chain fatty acids (SCFAs) were lower in MG subjects. Changes were also found in terms of serum biomarkers and fecal metabolites. A link was found between the bacterial Operational Taxonomic Unit (OTU), some metabolite biomarkers, and MG’s clinical symptoms. There were also variations in microbial and metabolic markers, which, in combination, could be used as an MG diagnostic tool, and interventions via fecal microbiota transplant (FMT) could affect MG development. Probiotics may influence MG by restoring the gut microbiome imbalance, aiding the prevention of MG, and lowering the risk of gut inflammation by normalizing serum biomarkers. Hence, this review will discuss how alterations of gut microbiome composition and function relate to MG and the benefits of gut modulation.
Collapse
|
11
|
Escherichia coli Nissle 1917 Enhances Efficacy of Oral Attenuated Human Rotavirus Vaccine in a Gnotobiotic Piglet Model. Vaccines (Basel) 2022; 10:vaccines10010083. [PMID: 35062744 PMCID: PMC8779073 DOI: 10.3390/vaccines10010083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 12/24/2022] Open
Abstract
Human rotavirus (HRV) infection is a major cause of viral gastroenteritis in young children worldwide. Current oral vaccines perform poorly in developing countries where efficacious vaccines are needed the most. Therefore, an alternative affordable strategy to enhance efficacy of the current RV vaccines is necessary. This study evaluated the effects of colonization of neonatal gnotobiotic (Gn) pigs with Escherichia coli Nissle (EcN) 1917 and Lacticaseibacillus rhamnosus GG (LGG) probiotics on immunogenicity and protective efficacy of oral attenuated (Att) HRV vaccine. EcN-colonized pigs had reduced virulent HRV (VirHRV) shedding and decreased diarrhea severity compared with the LGG-colonized group. They also had enhanced HRV-specific IgA antibody titers in serum and antibody secreting cell numbers in tissues pre/post VirHRV challenge, HRV-specific IgA antibody titers in intestinal contents, and B-cell subpopulations in tissues post VirHRV challenge. EcN colonization also enhanced T-cell immune response, promoted dendritic cells and NK cell function, reduced production of proinflammatory cytokines/Toll like receptor (TLR), and increased production of immunoregulatory cytokines/TLR expression in various tissues pre/post VirHRV challenge. Thus, EcN probiotic adjuvant with AttHRV vaccine enhances the immunogenicity and protective efficacy of AttHRV to a greater extent than LGG and it can be used as a safe and economical oral vaccine adjuvant.
Collapse
|
12
|
Investigation of Immunomodulatory and Gut Microbiota-Altering Properties of Multicomponent Nutraceutical Prepared from Lactic Acid Bacteria, Bovine Colostrum, Apple Production By-Products and Essential Oils. Foods 2021; 10:foods10061313. [PMID: 34200426 PMCID: PMC8229151 DOI: 10.3390/foods10061313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 01/10/2023] Open
Abstract
Dietary components, such as lactic acid bacteria (LAB), bovine colostrum, apple production by-products, and essential oils, can favorably alter the host immune system and gut microbiota, however, their cumulative effect as multicomponent nutraceutical supplement has not been investigated. Therefore, the present study is the first one to evaluate a combination of LAB, bovine colostrum, dehydrated apple pomace, and essential oils for their immunomodulatory and prebiotic properties in the swine model. This study shows that supplementary feeding of pigs using multicomponent nutraceutical resulted in a statistically significant decrease in proportions of T cytotoxic and double-positive (CD4+CD8+low) cells within the CD3+ cell population at 28 DPI, compared to the beginning of the experiment (0DPI). Conversely, a statistically significant increase in proportions of B cells (accompanied by an increase in IgG concentration) and macrophage/monocyte cells within viable cell population at 28 DPI, compared to the beginning of the experiments, was observed. Furthermore, changes in the bacterial composition of gut microbiota in pigs fed with multicomponent nutraceutical changed significantly, with a 1.78 times higher number of probiotic strains (Bifidobacterium, Streptococcus, Faecilbacterium) at the end of the experiment, compared to control group animals. This study shows a positive effect of the nutraceutical formula used on the changes of gut microbiota by facilitating an increase in probiotic bacteria strains and possible anti-inflammatory properties.
Collapse
|
13
|
Escherichia coli Nissle 1917 Enhances Innate and Adaptive Immune Responses in a Ciprofloxacin-Treated Defined-Microbiota Piglet Model of Human Rotavirus Infection. mSphere 2021; 6:6/2/e00074-21. [PMID: 33789939 PMCID: PMC8546683 DOI: 10.1128/msphere.00074-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human rotavirus (HRV) infection is a major cause of gastroenteritis in children worldwide. Broad-spectrum antibiotic-induced intestinal microbial imbalance and the ensuing immune-metabolic dysregulation contribute to the persistence of HRV diarrhea. Escherichia coli Nissle 1917 (EcN), a Gram-negative probiotic, was shown to be a potent immunostimulant and alleviated HRV-induced diarrhea in monocolonized gnotobiotic (Gn) piglets. Our goal was to determine how EcN modulates immune responses in ciprofloxacin (Cipro)-treated Gn piglets colonized with a defined commensal microbiota (DM) and challenged with virulent HRV (VirHRV). Cipro given in therapeutic doses for a short term reduced serum and intestinal total and HRV-specific antibody titers, while EcN treatment alleviated this effect. Similarly, EcN treatment increased the numbers of total immunoglobulin-secreting cells, HRV-specific antibody-secreting cells, activated antibody-forming cells, resting/memory antibody-forming B cells, and naive antibody-forming B cells in systemic and/or intestinal tissues. Decreased levels of proinflammatory but increased levels of immunoregulatory cytokines and increased frequencies of Toll-like receptor-expressing cells were evident in the EcN-treated VirHRV-challenged group. Moreover, EcN treatment increased the frequencies of T helper and T cytotoxic cells in systemic and/or intestinal tissues pre-VirHRV challenge and the frequencies of T helper cells, T cytotoxic cells, effector T cells, and T regulatory cells in systemic and/or intestinal tissues postchallenge. Moreover, EcN treatment increased the frequencies of systemic and mucosal conventional and plasmacytoid dendritic cells, respectively, and the frequencies of systemic natural killer cells. Our findings demonstrated that Cipro use altered immune responses of DM-colonized neonatal Gn pigs, while EcN supplementation rescued these immune parameters partially or completely. IMPORTANCE Rotavirus (RV) is a primary cause of malabsorptive diarrhea in children and is associated with significant morbidity and mortality, especially in developing countries. The use of antibiotics exacerbates intestinal microbial imbalance and results in the persistence of RV-induced diarrhea. Probiotics are now being used to treat enteric infections and ulcerative colitis. We showed previously that probiotics partially protected gnotobiotic (Gn) piglets against human RV (HRV) infection and decreased the severity of diarrhea by modulating immune responses. However, the interactions between antibiotic and probiotic treatments and HRV infection in the context of an established gut microbiota are poorly understood. In this study, we developed a Gn pig model to study antibiotic-probiotic-HRV interactions in the context of a defined commensal microbiota (DM) that mimics aspects of the infant gut microbiota. Our results provide valuable information that will contribute to the treatment of antibiotic- and/or HRV-induced diarrhea and may be applicable to other enteric infections in children.
Collapse
|
14
|
Patil Y, Gooneratne R, Ju XH. Interactions between host and gut microbiota in domestic pigs: a review. Gut Microbes 2020; 11:310-334. [PMID: 31760878 PMCID: PMC7524349 DOI: 10.1080/19490976.2019.1690363] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/16/2019] [Accepted: 11/04/2019] [Indexed: 02/03/2023] Open
Abstract
It is well established that pig gut microbiota plays a critical role in maintaining metabolic homeostasis as well as in a myriad of physiological, neurological and immunological functions; including protection from pathogens and digestion of food materials - some of which would be otherwise indigestible by the pig. A rich and diverse gut microbial ecosystem (balanced microbiota) is the hallmark of good health; while qualitative and quantitative perturbations in the microbial composition can lead to development of various diseases. Alternatively, diseases caused by stressors or other factors have been shown to negatively impact the microbiota. This review focuses primarily on how commensal microorganisms in the gastrointestinal tract of pigs influence biochemical, physiological, immunological, and metabolic processes within the host animal.
Collapse
Affiliation(s)
- Yadnyavalkya Patil
- College of Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, China
- Faculty of Agriculture and Life Sciences, Department of Wine, Food, and Molecular Biosciences, Lincoln University, Lincoln, Canterbury, New Zealand
- Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Ravi Gooneratne
- Faculty of Agriculture and Life Sciences, Department of Wine, Food, and Molecular Biosciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - Xiang-Hong Ju
- College of Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, China
- Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| |
Collapse
|
15
|
Hussein EOS, Ahmed SH, Abudabos AM, Aljumaah MR, Alkhlulaifi MM, Nassan MA, Suliman GM, Naiel MAE, Swelum AA. Effect of Antibiotic, Phytobiotic and Probiotic Supplementation on Growth, Blood Indices and Intestine Health in Broiler Chicks Challenged with Clostridium perfringens. Animals (Basel) 2020; 10:ani10030507. [PMID: 32197455 PMCID: PMC7142990 DOI: 10.3390/ani10030507] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Necrotic enteritis is one of the most important economic issues in the poultry industry, associated with sudden death rates of up to 50%. However, there is limited information on the role of probiotics and/or phytobiotic compounds on the treatment and prevention of Clostridium perfringens infections in broiler chicks. This study aimed to assess the effects of probiotic compounds (Maxus, CloStat, Sangrovit Extra, CloStat + Sangrovit Extra and Gallipro Tech) on the growth performance, blood biochemistry and intestinal health of broiler chicks in vivo. The results demonstrated that the inclusion of probiotic and/or phytobiotic compounds has a positive effect on performance, blood constituents, liver histopathology, intestinal morphology and histopathology. Furthermore, a notable reduction in both lesion scores was observed when probiotics and phytobiotics alone or in combination were included in the diets. Abstract This study evaluated the effects of feed additives on the growth, blood biochemistry and intestinal health of broiler chicks. A total of 378 of broiler chicks (Ross 308) were randomly allotted to seven groups. Chicks were fed a basal diet with 0.0 (control negative), 0.0 (control positive), 0.1, 0.5, 0.12, 0.5 + 0.12 and 0.2 g Kg−1 of Maxus, CloStat, Sangrovit Extra, CloStat + Sangrovit Extra and Gallipro Tech, respectively for 35 days. After 15 days, the chicks were inoculated with Clostridium perfringens. All feed additives were found to enhance growth performance and feed efficiency. The best feed conversion ratio was found in the Negative Control, CloStat + Sangrovit Extra and Gallipro Tect groups, respectively. A notable increase in villus length, total villus area, small intestine weight, ilium weight and total lesion score was found in chicks supplemented with Bacillus subtilis. Besides, the dietary inclusion of phytobiotic compounds showed potential in reducing the serum Alanine aminotransferase (ALT) concentration and increasing the glucose levels. All intestine and liver histopathological signs were reduced in chicks fed a probiotic-supplemented diet. Our findings indicate that supplementation with probiotics and phytobiotics alone or in combined form can be used to enhance performance, intestine health and blood constituents against C. perfringens infection in broiler chicks.
Collapse
Affiliation(s)
- Elsayed O. S. Hussein
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (G.M.S.); (A.A.S.)
- Correspondence: (E.O.S.H.); (A.M.A.)
| | - Shamseldein H. Ahmed
- Department of Basic Sciences, College of Veterinary Medicine, Sudan University of Science and Technology, P.O. Box 121, Khartoum 11123, Sudan;
| | - Alaeldein M. Abudabos
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (G.M.S.); (A.A.S.)
- Correspondence: (E.O.S.H.); (A.M.A.)
| | - Mashael R. Aljumaah
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.A.); (M.M.A.)
| | - Manal M. Alkhlulaifi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.A.); (M.M.A.)
| | - Mohamed A. Nassan
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt;
| | - Gamaleldin M. Suliman
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (G.M.S.); (A.A.S.)
| | - Mohammed A. E. Naiel
- Department of Animal Production, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt;
| | - Ayman A. Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (G.M.S.); (A.A.S.)
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
16
|
Michael H, Langel SN, Miyazaki A, Paim FC, Chepngeno J, Alhamo MA, Fischer DD, Srivastava V, Kathayat D, Deblais L, Rajashekara G, Saif LJ, Vlasova AN. Malnutrition Decreases Antibody Secreting Cell Numbers Induced by an Oral Attenuated Human Rotavirus Vaccine in a Human Infant Fecal Microbiota Transplanted Gnotobiotic Pig Model. Front Immunol 2020; 11:196. [PMID: 32117313 PMCID: PMC7033455 DOI: 10.3389/fimmu.2020.00196] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/24/2020] [Indexed: 01/31/2023] Open
Abstract
Human rotavirus (HRV) is a leading cause of morbidity and mortality in children, especially in developing countries. Malnutrition is prevalent in these countries, which may contribute to the decreased oral vaccine efficacy, posing a concern for global health. Neonatal gnotobiotic (Gn) pigs closely resemble human infants in their anatomy, physiology, and outbred status and are a unique model to investigate malnutrition, oral live attenuated HRV (AttHRV) vaccination, and subsequent virulent HRV (VirHRV) challenge. We evaluated the impact of malnutrition on AttHRV vaccine efficacy and B cell immune responses in neonatal germfree (GF) or Gn pigs transplanted with human infant fecal microbiota (HIFM). Pigs were fed either deficient or sufficient bovine milk diets. Malnutrition did not significantly affect the serum and intestinal contents total or HRV-specific IgG and IgA antibody titers pre VirHRV challenge. However, HRV-specific IgG and IgA antibody secreting cells (ASCs) were reduced in blood or intestinal tissues following AttHRV vaccination and pre VirHRV challenge in deficient HIFM transplanted pigs. Furthermore, post-VirHRV challenge, deficient HIFM pigs had decreased total Ig and HRV-specific IgG and IgA antibody titers in serum or intestinal contents, in addition to decreased HRV-specific IgG and IgA ASCs in blood and ileum, compared with sufficient HIFM pigs. Our results indicate that deficient diet impairs B cell mucosal, and systemic immune responses following HRV vaccination, and challenge. The impaired immune responses contributed to the decreased protective efficacy of the AttHRV vaccine, suggesting that malnutrition may significantly reduce the effectiveness of oral HRV vaccines in children in developing countries.
Collapse
Affiliation(s)
- Husheem Michael
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Stephanie N Langel
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Ayako Miyazaki
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States.,Division of Viral Disease and Epidemiology, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Francine C Paim
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Juliet Chepngeno
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Moyasar A Alhamo
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - David D Fischer
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Vishal Srivastava
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Dipak Kathayat
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Loic Deblais
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Linda J Saif
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Anastasia N Vlasova
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| |
Collapse
|
17
|
Salek Farrokhi A, Darabi N, Yousefi B, Askandar RH, Shariati M, Eslami M. Is it true that gut microbiota is considered as panacea in cancer therapy? J Cell Physiol 2019; 234:14941-14950. [PMID: 30786013 DOI: 10.1002/jcp.28333] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/26/2019] [Accepted: 01/30/2019] [Indexed: 02/06/2023]
Abstract
Recent studies demonstrated that a combination of the gut microbiome has the vital effect on the efficacy of anticancer immune therapies. Regulatory effects of microbiota have been shown in different types of cancer therapies such as chemotherapy and immunotherapy. Immune-checkpoint-blocked therapies are the recent efficient cancer immunotherapy strategies. The target of immune-checkpoint blocking is cytotoxic T lymphocyte protein-4 (CTLA-4) or blockade of programmed death-1 (PD-1) protein and its ligand programmed death ligand 1 (PD-L1) that they have been considered as cancer immunotherapy in recent years. In the latest studies, it have been demonstrated that several gut bacteria such as Akkermansia muciniphila, Bifidobacterium spp., Faecalibacterium spp., and Bacteroides fragilis have the regulatory effects on PD-1, PD-L1, and CTLA-4 blocked anticancer therapy outcome.
Collapse
Affiliation(s)
- Amir Salek Farrokhi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Narges Darabi
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Rafee Habib Askandar
- Nursing Department, Halabja Technical Institute, Sulaimani Polytechnic University, Sulaimani, Iraq
| | - Mansoreh Shariati
- Faculty of Basic Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
18
|
Reddy YS, Srivalliputturu SB, Bharatraj DK. The effect of lead (Pb) exposure and iron (Fe) deficiency on intestinal lactobacilli, E. coli and yeast: A study in experimental rats. J Occup Health 2018; 60:475-484. [PMID: 30210097 PMCID: PMC6281633 DOI: 10.1539/joh.2017-0267-oa] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 07/11/2018] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE The current study investigated the additive effect of oral lead (Pb) exposure and dietary iron (Fe) deficiency on intestinal lactobacilli, E. coli, and yeast in SD rats. METHODS Weanling rats were fed on control diet (CD) or iron deficient diet (ID) for 4 weeks, followed by oral Pb exposure for another 4 weeks. Lead exposure was withdrawn for 2 weeks, and then resumed after 2 weeks. Blood samples were collected to determine haemoglobin (Hb), serum iron, blood Pb and δ-Aminolevulenic acid dehydratase (ALAD) activity. Fecal samples were collected to enumerate the lactobacilli, E. coli and yeast population on selective agar media and determine Pb levels. RESULTS Hb and serum Fe levels decreased significantly in iron deficient rats. Pb exposed rats had a significant increase in blood Pb levels and decreased ALAD activity. The lactobacilli population was significantly decreased (p<0.05) in ID rats compared to the CD group. Further, a significant decrease in the lactobacilli population was observed in Pb exposed rats irrespective of the dietary regimen. Upon withdrawal of Pb exposure, lactobacilli increased significantly in both the CD+Pb and ID+Pb groups, whereas re-exposure to Pb decreased lactobacilli population. The E. coli and yeast populations were inconsistent among both the ID and Pb exposed rats compared to controls. Fecal Pb levels increased significantly in Pb exposed rats irrespective of diet. CONCLUSION An additive effect of dietary Fe deficiency and oral Pb exposure resulted in greater reductions in the intestinal lactobacilli population compared to either treatment alone. In addition, transient withdrawal of Pb exposure led to improved lactobacilli population irrespective of Fe status.
Collapse
|
19
|
Kumar A, Vlasova AN, Deblais L, Huang HC, Wijeratne A, Kandasamy S, Fischer DD, Langel SN, Paim FC, Alhamo MA, Shao L, Saif LJ, Rajashekara G. Impact of nutrition and rotavirus infection on the infant gut microbiota in a humanized pig model. BMC Gastroenterol 2018; 18:93. [PMID: 29929472 PMCID: PMC6013989 DOI: 10.1186/s12876-018-0810-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 05/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human rotavirus (HRV) is a major cause of viral gastroenteritis in infants; particularly in developing countries where malnutrition is prevalent. Malnutrition perturbs the infant gut microbiota leading to sub-optimal functioning of the immune system and further predisposing infants to enteric infections. Therefore, we hypothesized that malnutrition exacerbates rotavirus disease severity in infants. METHODS In the present study, we used a neonatal germ free (GF) piglets transplanted with a two-month-old human infant's fecal microbiota (HIFM) on protein deficient and sufficient diets. We report the effects of malnourishment on the HRV infection and the HIFM pig microbiota in feces, intestinal and systemic tissues, using MiSeq 16S gene sequencing (V4-V5 region). RESULTS Microbiota analysis indicated that the HIFM transplantation resulted in a microbial composition in pigs similar to that of the original infant feces. This model was then used to understand the interconnections between microbiota diversity, diet, and HRV infection. Post HRV infection, HIFM pigs on the deficient diet had lower body weights, developed more severe diarrhea and increased virus shedding compared to HIFM pigs on sufficient diet. However, HRV induced diarrhea and shedding was more pronounced in non-colonized GF pigs compared to HIFM pigs on either sufficient or deficient diet, suggesting that the microbiota alone moderated HRV infection. HRV infected pigs on sufficient diet showed increased microbiota diversity in intestinal tissues; whereas, greater diversity was observed in systemic tissues of HRV infected pigs fed with deficient diet. CONCLUSIONS These results suggest that proper nourishment improves the microbiota quality in the intestines, alleviates HRV disease and lower probability of systemic translocation of potential opportunistic pathogens/pathobionts. In conclusion, our findings further support the role for microbiota and proper nutrition in limiting enteric diseases.
Collapse
Affiliation(s)
- Anand Kumar
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
- Present address: Group B-10: Biosecurity and Public Health, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM USA
| | - Anastasia N. Vlasova
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Loic Deblais
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Huang-Chi Huang
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Asela Wijeratne
- The Molecular and Cellular Imaging Center, The Ohio State University, Wooster, OH USA
| | - Sukumar Kandasamy
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - David D. Fischer
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Stephanie N. Langel
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Francine Chimelo Paim
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Moyasar A. Alhamo
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Lulu Shao
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
- Present address: Hillman Cancer Center, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260 USA
| | - Linda J. Saif
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Gireesh Rajashekara
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| |
Collapse
|
20
|
Magwira CA, Taylor MB. Composition of gut microbiota and its influence on the immunogenicity of oral rotavirus vaccines. Vaccine 2018; 36:3427-3433. [PMID: 29752022 DOI: 10.1016/j.vaccine.2018.04.091] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/09/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Abstract
The introduction of oral rotavirus vaccines (ORVVs) has led to a reduction in number of hospitalisations and deaths due to rotavirus (RV) infection. However, the efficacy of the vaccines has been varied with low-income countries showing significantly lower efficacy as compared to high-income countries. The reasons for the disparity are not fully understood but are thought to be multi-factorial. In this review article, we discuss the concept that the disparity in the efficacy of oral rotavirus vaccines between the higher and lower socio-economical countries could be due the nature of the bacteria that colonises and establishes in the gut early in life. We further discuss recent studies that has demonstrated significant correlations between the composition of the gut bacteria and the immunogenicity of oral vaccines, and their implications in the development of novel oral RV vaccines or redesigning the current ones for maximum impact.
Collapse
Affiliation(s)
- Cliff A Magwira
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, South Africa.
| | - Maureen B Taylor
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, South Africa; School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, South Africa
| |
Collapse
|
21
|
Wang Y, Ma R, Liu F, Lee SA, Zhang L. Modulation of Gut Microbiota: A Novel Paradigm of Enhancing the Efficacy of Programmed Death-1 and Programmed Death Ligand-1 Blockade Therapy. Front Immunol 2018; 9:374. [PMID: 29556232 PMCID: PMC5845387 DOI: 10.3389/fimmu.2018.00374] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/09/2018] [Indexed: 12/26/2022] Open
Abstract
Blockade of programmed death 1 (PD-1) protein and its ligand programmed death ligand 1 (PD-L1) has been used as cancer immunotherapy in recent years, with the blockade of PD-1 being more widely used than blockade of PD-L1. PD-1 and PD-L1 blockade therapy showed benefits in patients with various types of cancer; however, such beneficial effects were seen only in a subgroup of patients. Improving the efficacy of PD-1 and PD-L1 blockade therapy is clearly needed. In this review, we summarize the recent studies on the effects of gut microbiota on PD-1 and PD-L1 blockade and discuss the new perspectives on improving efficacy of PD-1 and PD-L1 blockade therapy in cancer treatment through modulating gut microbiota. We also discuss the possibility that chronic infections or inflammation may impact on PD-1 and PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Yiming Wang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Rena Ma
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Fang Liu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Seul A. Lee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
22
|
Hayashi RM, Lourenço MC, Kraieski AL, Araujo RB, Gonzalez-Esquerra R, Leonardecz E, da Cunha AF, Carazzolle MF, Monzani PS, Santin E. Effect of Feeding Bacillus subtilis Spores to Broilers Challenged with Salmonella enterica serovar Heidelberg Brazilian Strain UFPR1 on Performance, Immune Response, and Gut Health. Front Vet Sci 2018; 5:13. [PMID: 29487856 PMCID: PMC5816941 DOI: 10.3389/fvets.2018.00013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 01/22/2018] [Indexed: 12/14/2022] Open
Abstract
Salmonellosis is a poultry industry and public health concern worldwide. Recently, Salmonella enterica serovar Heidelberg (SH) has been reported in broilers in Brazil. The effect of feeding a blend of three strains of Bacillus subtilis (PRO) was studied in broilers orally challenged (107 CFU/chick) or not with a SH isolated in south of Brazil (UFPR1 strain). Twelve male Cobb 500 broilers per pen were randomly assigned to six treatments in a 3 × 2 factorial experiment where PRO was added at 0, 250, or 500 g/ton of broiler feed and fed to either SH-challenged (SH Control, SH + PRO 250, and SH + PRO 500) or non-challenged birds (Control, PRO 250, and PRO 500). Broiler performance, histologic alterations in intestinal morphology, Salmonella quantification and immune cells counts in liver (macrophages, T CD4+ and T CD8+) were analyzed. Changes in the intestinal microbiota of broilers were also studied by metagenomics for Control, SH Control, SH + PRO 250, and SH + PRO 500 only. Feeding PRO at 250 or 500 g/ton reduced SH counts and incidence in liver and cecum at 21 days of age. It was observed that PRO groups increased the macrophage mobilization to the liver in SH-challenged birds (P < 0.05) but reduced these cells in the liver of non-challenged birds, showing an interesting immune cell dynamics effect. PRO at 250 g/ton did not affect gut histology, but improved animal performance (P < 0.05) while PRO at 500/ton did not affect animal performance but increased histologic alteration related to activation of the defense response in the ileum in SH challenged birds compared to control birds (P < 0.05). SH + PRO 500 group presented a more diverse cecal microbiota (Shannon–Wiener index; P < 0.05) compared to Control and SH Control groups; while SH + PRO 250 had greater ileal richness (JackkNife index) compared to Control (P < 0.05). PRO was effective in reducing Salmonella colonization in liver and cecum when fed at 250 or 500 g/ton to broilers inoculated with SH strain UFPR1. PRO promotes positive alterations in performance (at 250 g/ton), immune modulatory effect in the gastrointestinal tract, SH reduction, and intestinal microbiota modulation.
Collapse
Affiliation(s)
- Ricardo Mitsuo Hayashi
- Laboratório de Microbiologia e Ornitopatologia, Universidade Federal do Paraná, Curitiba, Brazil
| | - Mariana Camargo Lourenço
- Laboratório de Microbiologia e Ornitopatologia, Universidade Federal do Paraná, Curitiba, Brazil
| | | | | | | | - Eduardo Leonardecz
- Laboratório de Bioquímica e Genética Aplicada, Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Anderson Ferreira da Cunha
- Laboratório de Bioquímica e Genética Aplicada, Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, Brazil
| | | | - Paulo Sérgio Monzani
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - Elizabeth Santin
- Laboratório de Microbiologia e Ornitopatologia, Universidade Federal do Paraná, Curitiba, Brazil
| |
Collapse
|
23
|
Nallagatla H, Hemalatha R, Kondapalli NB, Mohammed S. Impact of a novel synbiotic supplementation during gestation and lactation on immune responses in the Swiss albino mice offspring. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2017; 97:2453-2459. [PMID: 27696418 DOI: 10.1002/jsfa.8059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/14/2016] [Accepted: 09/25/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Prebiotics from various regularly consumed cereals and novel substrates are currently being utilised as functional foods. The aim of this study was to determine the effect of synbiotic, formulated with prebiotic extracted from natural resources like green gram (Vigna radiata) along with probiotic Lactobacillus rhamnosus GG (LGG) in modulating immune responses in the offspring when supplemented during gestation and lactation. RESULTS Synbiotic supplementation was effective in improving cell mediated immunity and humoral immunity among F0 dams. Among F1 pups (F1 Syn + and F1 Syn-), synbiotic supplementation showed significantly heightened (P < 0.05) splenocyte proliferation, increased interleukin-10, interferon gamma and interleukin-17 responses, leucocyte phagocytic ability and increased secretory-immunoglobulin A. However, four-fold increase in IgG titres to Hepatitis-B vaccine was observed only in those mice that were supplemented with synbiotic postweaning (F1 Syn+). CONCLUSION Synbiotic supplementation to pregnant dams affected the offspring's cellular and mucosal immunity favorably. However, IgG response to Hepatitis-B vaccine was influenced positively only when the supplementation was extended to the offsprings in the post weaning period. © 2016 Society of Chemical Industry.
Collapse
Affiliation(s)
- Himaja Nallagatla
- Department of Clinical Microbiology & Immunology, National Institute of Nutrition (ICMR), Jamai-Osmania, Hyderabad, 500 007, Telangana, India
| | - Rajkumar Hemalatha
- Department of Clinical Microbiology & Immunology, National Institute of Nutrition (ICMR), Jamai-Osmania, Hyderabad, 500 007, Telangana, India
| | - Narendra Babu Kondapalli
- Department of Clinical Microbiology & Immunology, National Institute of Nutrition (ICMR), Jamai-Osmania, Hyderabad, 500 007, Telangana, India
| | - Shujauddin Mohammed
- Department of Clinical Microbiology & Immunology, National Institute of Nutrition (ICMR), Jamai-Osmania, Hyderabad, 500 007, Telangana, India
| |
Collapse
|
24
|
Kobayashi H, Kanmani P, Ishizuka T, Miyazaki A, Soma J, Albarracin L, Suda Y, Nochi T, Aso H, Iwabuchi N, Xiao JZ, Saito T, Villena J, Kitazawa H. Development of an in vitro immunobiotic evaluation system against rotavirus infection in bovine intestinal epitheliocytes. Benef Microbes 2017; 8:309-321. [PMID: 28042704 DOI: 10.3920/bm2016.0155] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The bovine intestinal epithelial cell line (BIE cells) expresses the Toll-like receptor (TLR)3 and is able to mount an antiviral immune response after the stimulation with poly(I:C). In the present study, we aimed to further characterise the antiviral defence mechanisms in BIE cells by evaluating the innate immune response triggered by rotavirus (RV) infection. In addition, we attempted to determine whether immunobiotic bifidobacteria are able to confer protection of BIE cells against RV infection by beneficially modulating the antiviral immune response. RV OSU (porcine) and UK (bovine) effectively infected BIE cells, while a significant lower capacity to infect BIE cells was observed for human (Wa) and murine (EW) RV. We observed that viral infection in BIE cells triggered TLR3/RIG-I-mediated immune responses with activation of IRF3 and TRAF3, induction of interferon beta (IFN-β) and up-regulation of inflammatory cytokines. Our results also demonstrated that preventive treatments with Bifidobacterium infantis MCC12 or Bifidobacterium breve MCC1274 significantly reduced RV titres in infected BIE cells and differentially modulated the innate immune response. Of note, both strains significantly improved the production of the antiviral factor IFN-β in RV-infected BIE cells. In conclusion, this work provides comprehensive information on the antiviral immune response of BIE cells against RV, that can be further studied for the development of strategies aimed to improve antiviral defences in bovine intestinal epithelial cells. Our results also demonstrate that BIE cells could be used as a newly immunobiotic evaluation system against RV infection for application in the bovine host.
Collapse
Affiliation(s)
- H Kobayashi
- 1 Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan.,2 Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan
| | - P Kanmani
- 1 Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan.,2 Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan
| | - T Ishizuka
- 1 Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan
| | - A Miyazaki
- 3 Viral Disease and Epidemiology Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki 305-0856, Japan
| | - J Soma
- 4 Research and Development Section, Zen-noh Institute of Animal Health, Sakura, Chiba 285-0043, Japan
| | - L Albarracin
- 1 Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan.,5 Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELACONICET), Chacabuco 145, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | - Y Suda
- 6 Department of Food, Agriculture and Environment, Miyagi University, 2-2-1 Hatadate, Taihaku-ku, Sendai, Miyagi 982-0215 Japan
| | - T Nochi
- 7 Infection Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan.,8 Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan
| | - H Aso
- 2 Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan.,8 Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan
| | - N Iwabuchi
- 9 Food Ingredients Institute, Morinaga Milk Industry Co. Ltd., 5-Chome, Higashihara, 252-8583 Zama-City, Kanagawa, Japan
| | - J-Z Xiao
- 10 Next Generation Science Institute, Morinaga Milk Industry Co. Ltd., 5-Chome, Higashihara, 252-8583 Zama-City, Kanagawa, Japan
| | - T Saito
- 1 Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan
| | - J Villena
- 1 Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan.,5 Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELACONICET), Chacabuco 145, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | - H Kitazawa
- 1 Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan.,2 Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, 468-1, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-0845, Japan
| |
Collapse
|
25
|
Villena J, Vizoso-Pinto MG, Kitazawa H. Intestinal Innate Antiviral Immunity and Immunobiotics: Beneficial Effects against Rotavirus Infection. Front Immunol 2016; 7:563. [PMID: 27994593 PMCID: PMC5136547 DOI: 10.3389/fimmu.2016.00563] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/22/2016] [Indexed: 12/13/2022] Open
Abstract
The mucosal tissues of the gastrointestinal tract are the main portal entry of pathogens such as rotavirus (RV), which is a leading cause of death due to diarrhea among young children across the globe and a major cause of severe acute intestinal infection in livestock animals. The interactions between intestinal epithelial cells (IECs) and immune cells with RVs have been studied for several years, and now, it is known that the innate immune responses triggered by this virus can have both beneficial and detrimental effects for the host. It was demonstrated that natural RV infection in infants and experimental challenges in mice result in the intestinal activation of pattern recognition receptors (PRRs) such as toll-like receptor 3 (TLR3) and striking secretion of proinflammatory mediators that can lead to increased local tissue damage and immunopathology. Therefore, modulating desregulated intestinal immune responses triggered by PRRs activation are a significant promise for reducing the burden of RV diseases. The ability of immunoregulatory probiotic microorganisms (immunobiotics) to protect against intestinal infections, such as those caused by RVs, is among the oldest effects studied for these important group of beneficial microbes. In this review, we provide an update of the current status on the modulation of intestinal antiviral innate immunity by immunobiotics and their beneficial impact on RV infection. In addition, we describe the research of our group that demonstrated the capacity of immunobiotic strains to beneficially modulated TLR3-triggered immune response in IECs, reduce the disruption of intestinal homeostasis caused by intraepithelial lymphocytes, and improve the resistance to RV infections.
Collapse
Affiliation(s)
- Julio Villena
- Immunobiotics Research Group, Tucuman, Argentina; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina; Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Maria Guadalupe Vizoso-Pinto
- Immunobiotics Research Group, Tucuman, Argentina; Faculty of Medicine, INSIBIO (UNT-CONICET), National University of Tucuman, Tucuman, Argentina
| | - Haruki Kitazawa
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman, Argentina; Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan; Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
26
|
Li JT, Wei J, Guo HX, Han JB, Ye N, He HY, Yu TT, Wu YZ. Development of a human rotavirus induced diarrhea model in Chinese mini-pigs. World J Gastroenterol 2016; 22:7135-7145. [PMID: 27610023 PMCID: PMC4988310 DOI: 10.3748/wjg.v22.i31.7135] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/26/2016] [Accepted: 06/13/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish a new animal model for the research of human rotavirus (HRV) infection, its pathogenesis and immunity and evaluation of potential vaccines.
METHODS: 5-d, 30-d and 60-d-old Chinese mini-pigs, Guizhou and Bamma, were inoculated with a single oral dose of attenuated strain Wa, G1, G3 of HRV, and PBS (control), respectively, and fecal samples of pigs from 0 to 7 d post infection (DPI) were collected individually. Enzyme linked immunosorbent assay was used to detect HRV antigen in feces. The HRV was tested by real-time PCR (RT-PCR). The sections of the intestinal tissue were stained with hematoxylin and eosin to observe the morphologic variation by microscopy. Immunofluorescence was used to determine the HRV in intestinal tissue. HRV particles in cells of the ileum were observed by electron micrography.
RESULTS: When inoculated with HRV, mini-pigs younger than 30 d developed diarrhea in an age-dependent manner and shed HRV antigen of the same inoculum, as demonstrated by RT-PCR. Histopathological changes were observed in HRV inoculated mini-pigs including small intestinal cell tumefaction and necrosis. HRV that was distributed in the small intestine was restricted to the top part of the villi on the internal wall of the ileum, which was observed by immunofluorescence and transmission electron microscopy. Virus particles were observed in Golgi like follicles in HRV-infected neonatal mini-pigs. Guizhou mini-pigs were more sensitive to HRV than Bamma with respect to RV antigen shedding and clinical diarrhea.
CONCLUSION: These results indicate that we have established a mini-pig model of HRV induced diarrhea. Our findings are useful for the understanding of the pathogenic mechanisms of HRV infection.
Collapse
|
27
|
Vlasova AN, Kandasamy S, Chattha KS, Rajashekara G, Saif LJ. Comparison of probiotic lactobacilli and bifidobacteria effects, immune responses and rotavirus vaccines and infection in different host species. Vet Immunol Immunopathol 2016; 172:72-84. [PMID: 26809484 DOI: 10.1016/j.vetimm.2016.01.003] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 01/12/2016] [Indexed: 02/06/2023]
Abstract
Different probiotic strains of Lactobacillus and Bifidobacterium genera possess significant and widely acknowledged health-promoting and immunomodulatory properties. They also provide an affordable means for prevention and treatment of various infectious, allergic and inflammatory conditions as demonstrated in numerous human and animal studies. Despite the ample evidence of protective effects of these probiotics against rotavirus (RV) infection and disease, the precise immune mechanisms of this protection remain largely undefined, because of limited mechanistic research possible in humans and investigated in the majority of animal models. Additionally, while most human clinical probiotic trials are well-standardized using the same strains, uniform dosages, regimens of the probiotic treatments and similar host age, animal studies often lack standardization, have variable experimental designs, and non-uniform and sometime limited selection of experimental variables or observational parameters. This review presents selected data on different probiotic strains of lactobacilli and bifidobacteria and summarizes the knowledge of their immunomodulatory properties and the associated protection against RV disease in diverse host species including neonates.
Collapse
Affiliation(s)
- Anastasia N Vlasova
- Food Animal Health Research Program, The Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, OH, USA.
| | - Sukumar Kandasamy
- Food Animal Health Research Program, The Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, OH, USA
| | - Kuldeep S Chattha
- Food Animal Health Research Program, The Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, OH, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, The Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, OH, USA
| | - Linda J Saif
- Food Animal Health Research Program, The Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, OH, USA.
| |
Collapse
|
28
|
Yang X, Twitchell E, Li G, Wen K, Weiss M, Kocher J, Lei S, Ramesh A, Ryan EP, Yuan L. High protective efficacy of rice bran against human rotavirus diarrhea via enhancing probiotic growth, gut barrier function, and innate immunity. Sci Rep 2015; 5:15004. [PMID: 26459937 PMCID: PMC4602212 DOI: 10.1038/srep15004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/11/2015] [Indexed: 12/16/2022] Open
Abstract
Previously, we showed that rice bran (RB) was able to reduce human rotavirus (HRV) diarrhea in gnotobiotic pigs. Here, we investigated its effect on the growth of diarrhea-reducing probiotic Lactobacillus rhamnosus GG (LGG) and Escherichia coli Nissle (EcN), and the resulting effects on HRV diarrhea, gut epithelial health, permeability and innate immune responses during virulent HRV challenge. On 3, 5, and 7 days of age pigs were inoculated with 2 × 104 colony-forming-units LGG+EcN to initiate colonization. Daily RB supplementation (replacing 10% calorie intake) was started at 5 days of age and continued until euthanasia. A subset of pigs in each group was challenged orally with 105 focus-forming-units of virulent HRV at 33 days of age. RB completely prevented HRV diarrhea in LGG+EcN colonized pigs. RB significantly promoted the growth of both probiotic strains in the gut (~5 logs) and increased the body-weight-gain at 4–5 weeks of age compared to non-RB group. After HRV challenge, RB-fed pigs had significantly lower ileal mitotic index and villus width, and significantly increased intestinal IFN-γ and total IgA levels compared to non-RB group. Therefore, RB plus LGG+EcN colonization may represent a highly effective therapeutic approach against HRV and potentially a variety of other diarrhea-inducing enteric pathogens.
Collapse
Affiliation(s)
- Xingdong Yang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Erica Twitchell
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Guohua Li
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Ke Wen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Mariah Weiss
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Jacob Kocher
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Shaohua Lei
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Ashwin Ramesh
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| |
Collapse
|
29
|
Lactobacillus rhamnosus GG Dosage Affects the Adjuvanticity and Protection Against Rotavirus Diarrhea in Gnotobiotic Pigs. J Pediatr Gastroenterol Nutr 2015; 60:834-43. [PMID: 25564808 DOI: 10.1097/mpg.0000000000000694] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVES The use of immunostimulatory strains of probiotics as adjuvants has been increasingly recognized as a promising approach in enhancing vaccine immunogenicity; however, dose effects of probiotic adjuvants are not well defined. In the present study, we examined dose effects of a commonly used probiotic strain, Lactobacillus rhamnosus GG (LGG), on immunomodulation with 2 different dosages. METHODS Neonatal gnotobiotic pigs were inoculated with 2 oral doses of attenuated human rotavirus (AttHRV) vaccines and fed with 5 doses (LGG5X; total 2.1 × 10(6) colony-forming units) or 9 doses (LGG9X; total 3.2 × 10(6) colony-forming units) of LGG, starting at 3 days of age. RESULTS Both LGG feeding regimens enhanced the protection rate of AttHRV vaccine against diarrhea on virulent human rotavirus challenge. LGG5X, but not LGG9X, significantly enhanced rotavirus-specific intestinal memory B-cell responses to AttHRV; LGG5X also significantly enhanced virus-specific intestinal immunoglobulin A (IgA) antibody-secreting cell responses. Both regimens significantly enhanced rotavirus-specific serum IgA antibody responses to AttHRV. They also enhanced rotavirus-specific interferon-γ-producing effector/memory T-cell responses to AttHRV vaccine, with LGG9X being more effective than LGG5X, and both regimens downregulated CD4+CD25-FoxP3+ regulatory T (Treg) cell responses in most lymphoid tissues examined prechallenge and postchallenge and maintained the CD4+CD25+FoxP3+ Treg population in the ileum and intraepithelial lymphocyte postchallenge. LGG9X, however, did not significantly reduce total CD4+CD25-FoxP3+ Treg frequencies in the intestine and transforming growth factor-β-producing and interleukin (IL)-10-producing Treg frequencies in the blood. CONCLUSIONS These results indicate that LGG at both dosages functioned as effective probiotic adjuvant for AttHRV vaccine, but different dosages differentially modulated immune responses to favor either the mucosal IgA response (LGG5X) or the T-cell response (LGG9X).
Collapse
|
30
|
Summerfield A, Auray G, Ricklin M. Comparative Dendritic Cell Biology of Veterinary Mammals. Annu Rev Anim Biosci 2015; 3:533-57. [DOI: 10.1146/annurev-animal-022114-111009] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Artur Summerfield
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland;
| | - Gael Auray
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland;
| | - Meret Ricklin
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland;
| |
Collapse
|
31
|
Effects of Lactobacillus acidophilus dietary supplementation on the performance, intestinal barrier function, rectal microflora and serum immune function in weaned piglets challenged with Escherichia coli lipopolysaccharide. Antonie van Leeuwenhoek 2015; 107:883-91. [DOI: 10.1007/s10482-015-0380-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/07/2015] [Indexed: 10/24/2022]
|
32
|
Hodgins DC, Chattha K, Vlasova A, Parreño V, Corbeil LB, Renukaradhya GJ, Saif LJ. Mucosal Veterinary Vaccines. Mucosal Immunol 2015. [PMCID: PMC7149859 DOI: 10.1016/b978-0-12-415847-4.00068-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Abstract
Enteric viral infections in domestic animals cause significant economic losses. The recent emergence of virulent enteric coronaviruses [porcine epidemic diarrhea virus (PEDV)] in North America and Asia, for which no vaccines are available, remains a challenge for the global swine industry. Vaccination strategies against rotavirus and coronavirus (transmissible gastroenteritis virus) infections are reviewed. These vaccination principles are applicable against emerging enteric infections such as PEDV. Maternal vaccines to induce lactogenic immunity, and their transmission to suckling neonates via colostrum and milk, are critical for early passive protection. Subsequently, in weaned animals, oral vaccines incorporating novel mucosal adjuvants (e.g., vitamin A, probiotics) may provide active protection when maternal immunity wanes. Understanding intestinal and systemic immune responses to experimental rotavirus and transmissible gastroenteritis virus vaccines and infection in pigs provides a basis and model for the development of safe and effective vaccines for young animals and children against established and emerging enteric infections.
Collapse
Affiliation(s)
- Kuldeep S Chattha
- Canadian Food Inspection Agency, Lethbridge, Alberta T1H 6P7, Canada;
| | | | | |
Collapse
|
34
|
Wen K, Tin C, Wang H, Yang X, Li G, Giri-Rachman E, Kocher J, Bui T, Clark-Deener S, Yuan L. Probiotic Lactobacillus rhamnosus GG enhanced Th1 cellular immunity but did not affect antibody responses in a human gut microbiota transplanted neonatal gnotobiotic pig model. PLoS One 2014; 9:e94504. [PMID: 24722168 PMCID: PMC3983166 DOI: 10.1371/journal.pone.0094504] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/16/2014] [Indexed: 12/16/2022] Open
Abstract
This study aims to establish a human gut microbiota (HGM) transplanted gnotobiotic (Gn) pig model of human rotavirus (HRV) infection and diarrhea, and to verify the dose-effects of probiotics on HRV vaccine-induced immune responses. Our previous studies using the Gn pig model found that probiotics dose-dependently regulated both T cell and B cell immune responses induced by rotavirus vaccines. We generated the HGM transplanted neonatal Gn pigs through daily feeding of neonatal human fecal suspension to germ-free pigs for 3 days starting at 12 hours after birth. We found that attenuated HRV (AttHRV) vaccination conferred similar overall protection against rotavirus diarrhea and virus shedding in Gn pigs and HGM transplanted Gn pigs. HGM promoted the development of the neonatal immune system, as evidenced by the significantly enhanced IFN-γ producing T cell responses and reduction of regulatory T cells and their cytokine production in the AttHRV-vaccinated pigs. The higher dose Lactobacillus rhamnosus GG (LGG) feeding (14 doses, up to 109 colony-forming-unit [CFU]/dose) effectively increased the LGG counts in the HGM Gn pig intestinal contents and significantly enhanced HRV-specific IFN-γ producing T cell responses to the AttHRV vaccine. Lower dose LGG (9 doses, up to 106 CFU/dose) was ineffective. Neither doses of LGG significantly improved the protection rate, HRV-specific IgA and IgG antibody titers in serum, or IgA antibody titers in intestinal contents compared to the AttHRV vaccine alone, suggesting that an even higher dose of LGG is needed to overcome the influence of the microbiota to achieve the immunostimulatory effect in the HGM pigs. This study demonstrated that HGM Gn pig is an applicable animal model for studying immune responses to rotavirus vaccines and can be used for studying interventions (i.e., probiotics and prebiotics) that may enhance the immunogenicity and protective efficacy of vaccines through improving the gut microbiota.
Collapse
Affiliation(s)
- Ke Wen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Christine Tin
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Haifeng Wang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Xingdong Yang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Guohua Li
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Ernawati Giri-Rachman
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Jacob Kocher
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Tammy Bui
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Sherrie Clark-Deener
- Department of Large Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Viginia, United States of America
| |
Collapse
|
35
|
Rubio-del-Campo A, Coll-Marqués JM, Yebra MJ, Buesa J, Pérez-Martínez G, Monedero V, Rodríguez-Díaz J. Noroviral p-particles as an in vitro model to assess the interactions of noroviruses with probiotics. PLoS One 2014; 9:e89586. [PMID: 24586892 PMCID: PMC3931819 DOI: 10.1371/journal.pone.0089586] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 01/22/2014] [Indexed: 11/19/2022] Open
Abstract
Noroviruses (NoVs) are the main etiologic agents of acute epidemic gastroenteritis and probiotic bacteria have been reported to exert a positive effect on viral diarrhea. The protruding (P) domain from NoVs VP1 capsid protein has the ability to assemble into the so-called P-particles, which retain the binding ability to host receptors. We purified the P-domains from NoVs genotypes GI.1 and GII.4 as 6X(His)-tagged proteins and determined that, similar to native domains, they were structured into P-particles that were functional in the recognition of the specific glycoconjugated receptors, as established by surface plasmon resonance experiments. We showed that several lactic acid bacteria (probiotic and non-probiotic) and a Gram-negative probiotic strain have the ability to bind P-particles on their surfaces irrespective of their probiotic status. The binding of P-particles (GI.1) to HT-29 cells in the presence of selected strains showed that bacteria can inhibit P-particle attachment in competitive exclusion experiments. However, pre-treatment of cells with bacteria or adding bacteria to cells with already attached P-particles enhanced the retention of the particles. Although direct viral binding and blocking of viral receptors have been postulated as mechanisms of protection against viral infection by probiotic bacteria, these results highlight the need for a careful evaluation of this hypothesis. The work presented here investigates for the first time the probiotic-NoVs-host interactions and points up the NoVs P-particles as useful tools to overcome the absence of in vitro cellular models to propagate these viruses.
Collapse
Affiliation(s)
- Antonio Rubio-del-Campo
- Laboratory of Lactic Acid Bacteria and Probiotics, Biotechnology Department, Institute of Agrochemistry and Food Technology (IATA-CSIC), Valencia, Spain
| | - José M. Coll-Marqués
- Laboratory of Lactic Acid Bacteria and Probiotics, Biotechnology Department, Institute of Agrochemistry and Food Technology (IATA-CSIC), Valencia, Spain
| | - María J. Yebra
- Laboratory of Lactic Acid Bacteria and Probiotics, Biotechnology Department, Institute of Agrochemistry and Food Technology (IATA-CSIC), Valencia, Spain
| | - Javier Buesa
- Department of Microbiology and Ecology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Gaspar Pérez-Martínez
- Laboratory of Lactic Acid Bacteria and Probiotics, Biotechnology Department, Institute of Agrochemistry and Food Technology (IATA-CSIC), Valencia, Spain
| | - Vicente Monedero
- Laboratory of Lactic Acid Bacteria and Probiotics, Biotechnology Department, Institute of Agrochemistry and Food Technology (IATA-CSIC), Valencia, Spain
- * E-mail: (VM); (JRD)
| | - Jesús Rodríguez-Díaz
- Laboratory of Lactic Acid Bacteria and Probiotics, Biotechnology Department, Institute of Agrochemistry and Food Technology (IATA-CSIC), Valencia, Spain
- * E-mail: (VM); (JRD)
| |
Collapse
|
36
|
Abstract
OBJECTIVE The aim of the study was to examine the dose effects of Lactobacillus acidophilus (LA) NCFM strain on rotavirus-specific antibody and B-cell responses in gnotobiotic pigs vaccinated with an oral attenuated human rotavirus (AttHRV). METHODS Pigs were inoculated with AttHRV vaccine in conjunction with high-dose LA (14 doses, total 2.2 × 10(6) colony-forming units [CFU]), intermediate-dose LA (MidLA) (9 doses, total 3.2 × 10(9) CFU), low-dose LA (LoLA) (5 doses, total 2.1 × 10(6) CFU), or without LA feeding. Protection against rotavirus shedding and diarrhea was assessed upon challenge with a virulent HRV. Rotavirus-specific immunoglobulin A (IgA) and IgG antibodies in serum and rotavirus-specific IgA and IgG antibody-secreting cells (ASCs) and memory B cells in ileum, spleen, and blood of the pigs were measured and compared among treatment groups. RESULTS The MidLA, but not high-dose LA or LoLA, significantly reduced rotavirus diarrhea (MidLA-only group) and significantly improved the protection conferred by AttHRV vaccine (MidLA + AttHRV group). Associated with the increased protection, MidLA significantly enhanced rotavirus-specific antibody, ASCs, and memory B-cell responses to AttHRV vaccine. High-dose LA or LoLA did not enhance virus-specific antibody and ASC responses, and hence did not improve the vaccine efficacy. CONCLUSIONS These findings highlight the importance of dose selection and indicate that certain specific lactobacilli strains at the appropriate dose have the dual function of reducing rotavirus diarrhea and enhancing the immunogenicity and protective efficacy of rotavirus vaccines.
Collapse
|
37
|
Vlasova AN, Chattha KS, Kandasamy S, Liu Z, Esseili M, Shao L, Rajashekara G, Saif LJ. Lactobacilli and bifidobacteria promote immune homeostasis by modulating innate immune responses to human rotavirus in neonatal gnotobiotic pigs. PLoS One 2013; 8:e76962. [PMID: 24098572 PMCID: PMC3788735 DOI: 10.1371/journal.pone.0076962] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/27/2013] [Indexed: 12/25/2022] Open
Abstract
The effects of co-colonization with Lactobacillus rhamnosus GG (LGG) and Bifidobacterium lactis Bb12 (Bb12) on 3-dose vaccination with attenuated HRV and challenge with virulent human rotavirus (VirHRV) were assessed in 4 groups of gnotobiotic (Gn) pigs: Pro+Vac (probiotic-colonized/vaccinated), Vac (vaccinated), Pro (probiotic-colonized, non-vaccinated) and Control (non-colonized, non-vaccinated). Subsets of pigs were euthanized pre- [post-challenge day (PCD) 0] and post (PCD7)-VirHRV challenge to assess diarrhea, fecal HRV shedding and dendritic cell/innate immune responses. Post-challenge, Pro+Vac and Vac groups were completely protected from diarrhea; protection rates against HRV shedding were 100% and 83%, respectively. Diarrhea and HRV shedding were reduced in Pro compared to Control pigs following VirHRV challenge. Diarrhea scores and virus shedding were significantly higher in Controls, compared to all other groups, coincident with significantly higher serum interferon-alpha levels post-challenge. LGG+Bb12 colonization ±vaccine promoted immunomaturation as reflected by increased frequencies of CD4, SWC3a, CD11R1, MHCII expressing mononuclear cells (MNCs) and conventional dendritic cells in intestinal tissues and blood post-challenge. Colonization decreased frequencies of toll-like receptors (TLR) 2 and TLR4 expressing MNCs from vaccinated pigs (Pro+Vac) pre-challenge and increased frequencies of TLR3 expressing MNCs from Pro pigs post-challenge, suggesting that probiotics likely exert anti-inflammatory (TLR2 and 4 down-regulation) and antiviral (TLR3 up-regulation by HRV dsRNA) actions via TLR signaling. Probiotic colonization alone (Pro) increased frequencies of intestinal and systemic apoptotic MNCs pre-challenge, thereby regulating immune hyperreactivity and tolerance. However, these frequencies were decreased in intestinal and systemic tissues post-challenge, moderating HRV-induced apoptosis. Additionally, post-challenge, Pro+Vac and Pro groups had significantly decreased MNC proliferation, suggesting that probiotics control excessive lymphoproliferative reactions upon VirHRV challenge. We conclude that in the neonatal Gn pig disease model, selected probiotics contribute to immunomaturation, regulate immune homeostasis and modulate vaccine and virulent HRV effects, thereby moderating HRV diarrhea.
Collapse
Affiliation(s)
- Anastasia N. Vlasova
- The Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, Ohio, United States of America
- * E-mail: (ANV); (LJS)
| | - Kuldeep S. Chattha
- The Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, Ohio, United States of America
| | - Sukumar Kandasamy
- The Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, Ohio, United States of America
| | - Zhe Liu
- The Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, Ohio, United States of America
| | - Malak Esseili
- The Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, Ohio, United States of America
| | - Lulu Shao
- The Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, Ohio, United States of America
| | - Gireesh Rajashekara
- The Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, Ohio, United States of America
| | - Linda J. Saif
- The Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, Ohio, United States of America
- * E-mail: (ANV); (LJS)
| |
Collapse
|
38
|
Wu S, Yuan L, Zhang Y, Liu F, Li G, Wen K, Kocher J, Yang X, Sun J. Probiotic Lactobacillus rhamnosus GG mono-association suppresses human rotavirus-induced autophagy in the gnotobiotic piglet intestine. Gut Pathog 2013; 5:22. [PMID: 23924832 PMCID: PMC3750464 DOI: 10.1186/1757-4749-5-22] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 07/25/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Human rotavirus (HRV) is the most important cause of severe diarrhea in infants and young children. Probiotic Lactobacillus rhamnosus GG (LGG) reduces rotavirus infection and diarrhea. However, the molecular mechanisms of LGG-mediated protection from rotavirus infection are poorly understood. Autophagy plays an essential role in responses to microbial pathogens. However, the role of autophagy in HRV infection and LGG treatment is unknown. We hypothesize that rotavirus gastroenteritis activates autophagy and that LGG suppresses virus-induced autophagy and prevents intestinal damage in infected piglets. METHODS We used LGG feeding to combat viral gastroenteritis in the gnotobiotic pig model of virulent HRV infection. RESULTS We found that LGG feeding did not increase autophagy, whereas virus infection induced autophagy in the piglet intestine. Virus infection increased the protein levels of the autophagy markers ATG16L1 and Beclin-1 and the autophagy regulator mTOR. LGG treatment during viral gastroenteritis reduced autophagy marker expression to normal levels, induced apoptosis and partially prevented virus-induced tissue damage. CONCLUSION Our study provides new insights into virus-induced autophagy and LGG suppression of uncontrolled autophagy and intestinal injury. A better understanding of the antiviral activity of LGG will lead to novel therapeutic strategies for infant infectious diseases.
Collapse
Affiliation(s)
- Shaoping Wu
- Department of Biochemistry, Rush University, Cohn Research Building, 1735 W. Harrison Street, Chicago, IL 60612, USA
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Integrated Life Science Building, 1981 Kraft Dr, Blacksburg, VA 24061-0913, USA
| | - Yongguo Zhang
- Department of Biochemistry, Rush University, Cohn Research Building, 1735 W. Harrison Street, Chicago, IL 60612, USA
| | - Fangning Liu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Integrated Life Science Building, 1981 Kraft Dr, Blacksburg, VA 24061-0913, USA
| | - Guohua Li
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Integrated Life Science Building, 1981 Kraft Dr, Blacksburg, VA 24061-0913, USA
| | - Ke Wen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Integrated Life Science Building, 1981 Kraft Dr, Blacksburg, VA 24061-0913, USA
| | - Jacob Kocher
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Integrated Life Science Building, 1981 Kraft Dr, Blacksburg, VA 24061-0913, USA
| | - Xingdong Yang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Integrated Life Science Building, 1981 Kraft Dr, Blacksburg, VA 24061-0913, USA
| | - Jun Sun
- Department of Biochemistry, Rush University, Cohn Research Building, 1735 W. Harrison Street, Chicago, IL 60612, USA
| |
Collapse
|
39
|
Abstract
Rotavirus is a leading cause of severe dehydrating diarrhoea in infants and young children. Following rotavirus infection in the intestine an innate immune response is rapidly triggered. This response leads to the induction of type I and type III interferons (IFNs) and other cytokines, resulting in a reduction in viral replication. Here we review the current literature describing the detection of rotavirus infection by pattern recognition receptors within host cells, the subsequent molecular mechanisms leading to IFN and cytokine production, and the processes leading to reduced rotavirus replication and the development of protective immunity. Rotavirus countermeasures against innate responses, and their roles in modulating rotavirus replication in mice, also are discussed. By linking these different aspects of innate immunity, we provide a comprehensive overview of the host’s first line of defence against rotavirus infection. Understanding these processes is expected to be of benefit in improving strategies to combat rotavirus disease.
Collapse
Affiliation(s)
- Gavan Holloway
- Department of Microbiology and Immunology, The University of Melbourne, Victoria 3010, Australia
| | - Barbara S. Coulson
- Department of Microbiology and Immunology, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
40
|
Auray G, Facci MR, van Kessel J, Buchanan R, Babiuk LA, Gerdts V. Porcine neonatal blood dendritic cells, but not monocytes, are more responsive to TLRs stimulation than their adult counterparts. PLoS One 2013; 8:e59629. [PMID: 23667422 PMCID: PMC3648567 DOI: 10.1371/journal.pone.0059629] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 02/15/2013] [Indexed: 01/20/2023] Open
Abstract
The neonatal immune system is often considered as immature or impaired compared to the adult immune system. This higher susceptibility to infections is partly due to the skewing of the neonatal immune response towards a Th2 response. Activation and maturation of dendritic cells (DCs) play an important role in shaping the immune response, therefore, DCs are a target of choice for the development of efficient and protective vaccine formulations able to redirect the neonatal immune response to a protective Th1 response. As pigs are becoming more important for vaccine development studies due to their similarity to the human immune system, we decided to compare the activation and maturation of a subpopulation of porcine DCs in adult and neonatal pigs following stimulation with different TLR ligands, which are promising candidates for adjuvants in vaccine formulations. Porcine blood derived DCs (BDCs) were directly isolated from blood and consisted of a mix of conventional and plasmacytoid DCs. Following CpG ODN (TLR9 ligand) and imiquimod (TLR7 ligand) stimulation, neonatal BDCs showed higher levels of expression of costimulatory molecules and similar (CpG ODN) or higher (imiquimod) levels of IL-12 compared to adult BDCs. Another interesting feature was that only neonatal BDCs produced IFN-α after TLR7 or TLR9 ligand stimulation. Stimulation with CpG ODN and imiquimod also induced enhanced expression of several chemokines. Moreover, in a mixed leukocyte reaction assay, neonatal BDCs displayed a greater ability to induce lymphoproliferation. These findings suggest that when stimulated via TLR7 or TLR9 porcine DCs display similar if not better response than adult porcine DCs.
Collapse
Affiliation(s)
- Gael Auray
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
- Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Marina R. Facci
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | - Jill van Kessel
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | - Rachelle Buchanan
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | | | - Volker Gerdts
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
- * E-mail:
| |
Collapse
|
41
|
Vlasova AN, Chattha KS, Kandasamy S, Siegismund CS, Saif LJ. Prenatally acquired vitamin A deficiency alters innate immune responses to human rotavirus in a gnotobiotic pig model. THE JOURNAL OF IMMUNOLOGY 2013; 190:4742-53. [PMID: 23536630 DOI: 10.4049/jimmunol.1203575] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We examined how prenatally acquired vitamin A deficiency (VAD) modulates innate immune responses and human rotavirus (HRV) vaccine efficacy in a gnotobiotic (Gn) piglet model of HRV diarrhea. The VAD and vitamin A-sufficient (VAS) Gn pigs were vaccinated with attenuated HRV (AttHRV) with or without concurrent oral vitamin A supplementation (100,000 IU) and challenged with virulent HRV (VirHRV). Regardless of vaccination status, the numbers of conventional and plasmacytoid dendritic cells (cDCs and pDCs) were higher in VAD piglets prechallenge, but decreased substantially postchallenge as compared with VAS pigs. We observed significantly higher frequency of CD103 (integrin αEβ7) expressing DCs in VAS versus VAD piglets postchallenge, indicating that VAD may interfere with homing (including intestinal) phenotype acquisition. Post-VirHRV challenge, we observed longer and more pronounced diarrhea and higher VirHRV fecal titers in nonvaccinated VAD piglets. Consistent with higher VirHRV shedding titers, higher IFN-α levels were induced in control VAD versus VAS piglet sera at postchallenge day 2. Ex vivo HRV-stimulated mononuclear cells (MNCs) isolated from spleen and blood of VAD pigs prechallenge also produced more IFN-α. In contrast, at postchallenge day 10, we observed reduced IFN-α levels in VAD pigs that coincided with decreased TLR3(+) MNC frequencies. Numbers of necrotic MNCs were higher in VAD pigs in spleen (coincident with splenomegaly in other VAD animals) prechallenge and intestinal tissues (coincident with higher VirHRV induced intestinal damage) postchallenge. Thus, prenatal VAD caused an imbalance in innate immune responses and exacerbated VirHRV infection, whereas vitamin A supplementation failed to compensate for these VAD effects.
Collapse
Affiliation(s)
- Anastasia N Vlasova
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Veterinary Preventive Medicine Department, The Ohio State University, Wooster, OH 44691, USA
| | | | | | | | | |
Collapse
|
42
|
Hemarajata P, Versalovic J. Effects of probiotics on gut microbiota: mechanisms of intestinal immunomodulation and neuromodulation. Therap Adv Gastroenterol 2013; 6:39-51. [PMID: 23320049 PMCID: PMC3539293 DOI: 10.1177/1756283x12459294] [Citation(s) in RCA: 623] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recent explorations of the human gut microbiota suggest that perturbations of microbial communities may increase predisposition to different disease phenotypes. Dietary nutrients may be converted into metabolites by intestinal microbes that serve as biologically active molecules affecting regulatory functions in the host. Probiotics may restore the composition of the gut microbiome and introduce beneficial functions to gut microbial communities, resulting in amelioration or prevention of gut inflammation and other intestinal or systemic disease phenotypes. This review describes how diet and intestinal luminal conversion by gut microbes play a role in shaping the structure and function of intestinal microbial communities. Proposed mechanisms of probiosis include alterations of composition and function of the human gut microbiome, and corresponding effects on immunity and neurobiology.
Collapse
Affiliation(s)
- Peera Hemarajata
- Department of Molecular Virology and Microbiology and Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children’s Hospital, 1102 Bates Street, Feigin Center 830, Houston, TX 77030, USA
| |
Collapse
|
43
|
Abstract
The delivery of certain living microorganisms in food has long been suggested as having positive health effects in humans. This practice has extended into food animal production, with a variety of microorganisms being used; lactic acid bacteria, various Bacillus species and the yeast Saccharomyces cerevisiae have been particularly used in the pig industry. The increased interest in probiotics is essentially due to the problem of microbial resistance to antibiotics and following the ban of the use of antibiotics in animal production, probiotics being considered an alternative means to reduce pathogen infection and improve animal health especially around the time of weaning. However, there is still a need to clarify the probiotic effectiveness in pigs, and the underlying mechanisms. When assessing the efficacy of probiotics one must consider the particular strain of organism being used and the production stage of the pigs being treated. The reproducible delivery of probiotics in industrial pig production is problematic as maintenance of viability is key to their beneficial activity, but difficult to achieve with commonly used feed processing technologies. One specific context where probiotics organisms may be reliably delivered is in systems utilising fermented liquid feeds. Liquid feed may be fermented by the activity of wild lactic acid bacteria or may be stimulated using specific isolates as 'starters'; the latter system has advantages in terms of reproducibility and speed of fermentation. The farm context in which the organism is used is likely to be critical; the use of probiotics is more likely to result in measurable economic gains in animals living in sub-optimal conditions rather than in those reared in the highest welfare and husbandry conditions. The establishment of a beneficial lactic acid bacteria population at birth may lead to healthier animals, this may be most effectively achieved by treating sows, which provide an amplification step and flood the neonatal pigs' environment with desirable bacterial strains. In contrast, it may be sufficient to provide a supportive, protective microbiota around the time of weaning as this is a time of major crisis with instability and loss of certain bacterial populations.
Collapse
|
44
|
Preidis GA, Saulnier DM, Blutt SE, Mistretta TA, Riehle KP, Major AM, Venable SF, Barrish JP, Finegold MJ, Petrosino JF, Guerrant RL, Conner ME, Versalovic J. Host response to probiotics determined by nutritional status of rotavirus-infected neonatal mice. J Pediatr Gastroenterol Nutr 2012; 55:299-307. [PMID: 22343914 PMCID: PMC4010314 DOI: 10.1097/mpg.0b013e31824d2548] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Beneficial microbes and probiotics are promising agents for the prevention and treatment of enteric and diarrheal diseases in children; however, little is known about their in vivo mechanisms of action. We used a neonatal mouse model of rotavirus diarrhea to gain insight into how probiotics ameliorate acute gastroenteritis. METHODS Rotavirus-infected mice were treated with 1 of 2 strains of human-derived Lactobacillus reuteri. We assessed intestinal microbiome composition with 16S metagenomic sequencing, enterocyte migration and proliferation with 5-bromo-2'-deoxyuridine, and antibody and cytokine concentrations with multiplex analyses of intestinal explant cultures. RESULTS Probiotics reduced diarrhea duration, improved intestinal histopathology, and enhanced intestinal microbiome richness and phylogenetic diversity. The magnitude of reduction of diarrhea by probiotics was strain specific and influenced by nutritional status. L reuteri DSM 17938 reduced diarrhea duration by 0, 1, and 2 days in underweight, normal weight, and overweight pups, respectively. The magnitude of reduction of diarrhea duration correlated with increased enterocyte proliferation and migration. Strain ATCC PTA 6475 reduced diarrhea duration by 1 day in all of the mice without increasing enterocyte proliferation. Both probiotic strains decreased concentrations of proinflammatory cytokines, including macrophage inflammatory protein-1α and interleukin-1β, in all of the animals, and increased rotavirus-specific antibodies in all but the underweight animals. Body weight also influenced the host response to rotavirus, in terms of diarrhea duration, enterocyte turnover, and antibody production. CONCLUSIONS These data suggest that probiotic enhancement of enterocyte proliferation, villus repopulation, and virus-specific antibodies may contribute to diarrhea resolution, and that nutritional status influences the host response to both beneficial microbes and pathogens.
Collapse
Affiliation(s)
- Geoffrey A. Preidis
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX
| | | | - Sarah E. Blutt
- the Department of Molecular Virology and Microbiology, Houston, TX
| | | | - Kevin P. Riehle
- the Bioinformatics Research Laboratory, Baylor College of Medicine, Houston, TX
| | - Angela M. Major
- the Department of Pathology, Texas Children's Hospital, Houston, TX
| | | | - James P. Barrish
- the Department of Pathology, Texas Children's Hospital, Houston, TX
| | | | - Joseph F. Petrosino
- the Department of Molecular Virology and Microbiology, Houston, TX
- the Human Genome Sequencing Center, Houston, TX
| | - Richard L. Guerrant
- the Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA
| | | | - James Versalovic
- the Department of Pathology, Texas Children's Hospital, Houston, TX
| |
Collapse
|
45
|
Hosoya S, Villena J, Chiba E, Shimazu T, Suda Y, Aso H, Saito T, Kitazawa H. Advanced application of porcine intestinal epithelial cells for the selection of immunobiotics modulating toll-like receptor 3-mediated inflammation. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2012; 46:474-81. [PMID: 22727542 DOI: 10.1016/j.jmii.2012.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 03/22/2012] [Accepted: 04/18/2012] [Indexed: 01/28/2023]
Abstract
PURPOSE In this study, we aimed to characterize toll-like receptor (TLR)-3-mediated inflammatory immune response in porcine intestinal epithelial (PIE) cells and in PIE-immune cell co-cultures and, to evaluate if these in vitro systems are useful for selecting immunomodulatory lactic acid bacteria. RESULTS We demonstrated that these systems are valuable tools for the in vitro study of the inflammatory response triggered by TLR3 in intestinal epithelial cells (IECs) and of the interaction between IECs and immune cells. In addition, we showed that PIE cells could be used for the selection of immunobiotic lactobacilli strains with anti-inflammatory activities. We found that Lactobacillus casei MEP221114 is an immunobiotic candidate for modulation of TLR3-mediated inflammatory responses. CONCLUSION The present study deepened our understanding of the mechanisms of immunobiotic action by demonstrating that the interaction between some lactobacilli strains and IECs can up-regulate the mRNA expression of TLR negative regulators and that this effect could help to regulate the production of inflammatory mediators during the generation of a TLR3-mediated immune response.
Collapse
Affiliation(s)
- Shoichi Hosoya
- Food Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Julio Villena
- Food Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Eriko Chiba
- Food Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Tomoyuki Shimazu
- Laboratory of Animal Breading and Genetics, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Yoshihito Suda
- Department of Food, Agriculture and Environmental Sciences, Miyagi University, Sendai 982-0215, Japan
| | - Hisashi Aso
- Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Tadao Saito
- Food Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Haruki Kitazawa
- Food Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan.
| |
Collapse
|
46
|
Azevedo MSP, Zhang W, Wen K, Gonzalez AM, Saif LJ, Yousef AE, Yuan L. Lactobacillus acidophilus and Lactobacillus reuteri modulate cytokine responses in gnotobiotic pigs infected with human rotavirus. Benef Microbes 2012; 3:33-42. [PMID: 22348907 DOI: 10.3920/bm2011.0041] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Probiotic lactic acid bacteria (LAB) have been shown to alleviate inflammation, enhance the immunogenicity of rotavirus vaccines, or reduce the severity of rotavirus diarrhoea. Although the mechanisms are not clear, the differential Th1/Th2/Th3-driving capacities and modulating effects on cytokine production of different LAB strains may be the key. Our goal was to delineate the influence of combining two probiotic strains of Lactobacillus acidophilus and Lactobacillus reuteri on the development of cytokine responses in neonatal gnotobiotic pigs infected with human rotavirus (HRV). We demonstrated that HRV alone, or HRV plus LAB, but not LAB alone, initiated serum cytokine responses, as indicated by significantly higher concentrations of IFN-α, IFN-γ, IL-12, and IL-10 at postinoculation day (PID) 2 in the HRV only and LAB+HRV+ pigs compared to LAB only and LAB-HRV- pigs. Peak cytokine responses coincided with the peak of HRV replication. LAB further enhanced the Th1 and Th2 cytokine responses to HRV infection as indicated by significantly higher concentrations of IL-12, IFN-γ, IL-4 and IL-10 in the LAB+HRV+ pigs compared to the LAB-HRV+ pigs. The LAB+HRV+ pigs maintained relatively constant concentrations of TGF-β compared to the HRV only group which had a significant increase at PID 2 and decrease at PID 7, suggesting a regulatory role of LAB in maintaining gut homeostasis. At PID 28, cytokine secreting cell (CSC) responses, measured by ELISpot, showed increased Th1 (IL-12, IFN-γ) CSC numbers in the LAB+HRV+ and LAB-HRV+ groups compared to LAB only and LAB-HRV- pigs, with significantly increased IL-12 CSCs in spleen and PBMCs and IFN-γ CSCs in spleen of the LAB+HRV+ group. Thus, HRV infection alone, but not LAB alone was effective in inducing cytokine responses but LAB significantly enhanced both Th1 and Th2 cytokines in HRV-infected pigs. LAB may also help to maintain immunological homeostasis during HRV infection by regulating TGF-β production.
Collapse
Affiliation(s)
- M S P Azevedo
- US Food and Drug Administration, National Center for Toxicological Research, Division of Microbiology, Jefferson, AR 72079, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Immunobiotic Lactobacillus jensenii modulates the Toll-like receptor 4-induced inflammatory response via negative regulation in porcine antigen-presenting cells. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1038-53. [PMID: 22573738 DOI: 10.1128/cvi.00199-12] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Previously, we demonstrated that Lactobacillus jensenii TL2937 attenuates the inflammatory response triggered by activation of Toll-like receptor 4 (TLR-4) in porcine intestinal epithelial cells. In view of the critical importance of antigen-presenting cell (APC) polarization in immunoregulation, the objective of the present study was to examine the effect of strain TL2937 on the activation patterns of APCs from swine Peyer's patches (PPs). We demonstrated that direct exposure of porcine APCs to L. jensenii in the absence of inflammatory signals increased expression of interleukin-10 (IL-10) and transforming growth factor β in CD172a(+) APCs and caused them to display tolerogenic properties. In addition, pretreatment of CD172a(+) APCs with L. jensenii resulted in differential modulation of the production of pro- and anti-inflammatory cytokines in response to TLR4 activation. The immunomodulatory effect of strain TL2937 was not related to a downregulation of TLR4 but was related to an upregulation of the expression of three negative regulators of TLRs: single immunoglobulin IL-1-related receptor (SIGIRR), A20, and interleukin-1 receptor-associated kinase M (IRAK-M). Our results also indicated that TLR2 has an important role in the anti-inflammatory activity of L. jensenii TL2937, since anti-TLR2 antibodies blocked the upregulation of SIGIRR and IRAK-M in CD172a(+) APCs and the production of IL-10 in response to TLR4 activation. We performed, for the first time, a precise functional characterization of porcine APCs from PPs, and we demonstrated that CD172a(+) cells were tolerogenic. Our findings demonstrate that adherent cells and isolated CD172a(+) cells harvested from swine PPs were useful for in vitro study of the inflammatory responses in the porcine gut and the immunomodulatory effects of immunobiotic microorganisms.
Collapse
|
48
|
Abstract
Keeping a delicate balance in the immune system by eliminating invading pathogens, while still maintaining self-tolerance to avoid autoimmunity, is critical for the body's health. The gut microbiota that resides in the gastrointestinal tract provides essential health benefits to its host, particularly by regulating immune homeostasis. Moreover, it has recently become obvious that alterations of these gut microbial communities can cause immune dysregulation, leading to autoimmune disorders. Here we review the advances in our understanding of how the gut microbiota regulates innate and adaptive immune homeostasis, which in turn can affect the development of not only intestinal but also systemic autoimmune diseases. Exploring the interaction of gut microbes and the host immune system will not only allow us to understand the pathogenesis of autoimmune diseases but will also provide us new foundations for the design of novel immuno- or microbe-based therapies.
Collapse
Affiliation(s)
- Hsin-Jung Wu
- Department of Immunobiology; College of Medicine; University of Arizona; Tucson, AZ USA,Arizona Arthritis Center; College of Medicine; University of Arizona; Tucson, AZ USA,Correspondence to: Hsin-Jung Wu;
| | - Eric Wu
- Department of Immunobiology; College of Medicine; University of Arizona; Tucson, AZ USA
| |
Collapse
|
49
|
Wen K, Li G, Bui T, Liu F, Li Y, Kocher J, Lin L, Yang X, Yuan L. High dose and low dose Lactobacillus acidophilus exerted differential immune modulating effects on T cell immune responses induced by an oral human rotavirus vaccine in gnotobiotic pigs. Vaccine 2011; 30:1198-207. [PMID: 22178726 DOI: 10.1016/j.vaccine.2011.11.107] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 11/28/2011] [Accepted: 11/29/2011] [Indexed: 01/14/2023]
Abstract
BACKGROUND Strain-specific effects of probiotics in pro- or anti-inflammatory immune responses have been well recognized. Several proinflammatory Lactobacillus strains have been shown to act as adjuvants to enhance the immunogenicity of vaccines. However, dose effects of probiotics in modulating immune responses are not clearly understood. This study examined the dose effects of Lactobacillus acidophilus (LA) NCFM strain on T cell immune responses to rotavirus vaccination in a gnotobiotic (Gn) pig model. METHODS Frequencies of IFN-γ producing CD4+ and CD8+ T cell and IL-10 and TGF-β producing CD4+CD25+ and CD4+CD25- regulatory T (Treg) cell responses were determined in the intestinal and systemic lymphoid tissues of Gn pigs vaccinated with an oral human rotavirus vaccine in conjunction with low dose (5 feedings; up to 10(6) colony forming units [CFU]/dose) or high dose (14 feedings; up to 10(9)CFU/dose) or without LA feeding. RESULTS Low dose LA significantly promoted IFN-γ producing T cell responses and down-regulated Treg cell responses and their TGF-β and IL-10 productions in all the tissues compared to the high dose LA and control groups. To the contrary, high dose LA increased the frequencies of Treg cells in most of the tissues compared to the control groups. The dose effects of LA on IFN-γ producing T cell and CD4+CD25- Treg cell immune responses were similar in the intestinal and systemic lymphoid tissues and were independent from the vaccination. CONCLUSION Thus the same probiotic strain in different doses can either promote or suppress IFN-γ producing T cell or Treg cell immune responses. These findings have significant implications in the use of probiotic lactobacilli as immunostimulatory versus immunoregulatory agents. Probiotics can be ineffective or even detrimental if not used at the optimal dosage for the appropriate purposes.
Collapse
Affiliation(s)
- Ke Wen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Integrated Life Science Building, 1981 Kraft Dr, Blacksburg, VA 24061-0913, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hosoya S, Villena J, Shimazu T, Tohno M, Fujie H, Chiba E, Shimosato T, Aso H, Suda Y, Kawai Y, Saito T, Alvarez S, Ikegami S, Itoh H, Kitazawa H. Immunobiotic lactic acid bacteria beneficially regulate immune response triggered by poly(I:C) in porcine intestinal epithelial cells. Vet Res 2011; 42:111. [PMID: 22046952 PMCID: PMC3220634 DOI: 10.1186/1297-9716-42-111] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 11/03/2011] [Indexed: 12/18/2022] Open
Abstract
This study analyzed the functional expression of TLR3 in various gastrointestinal tissues from adult swine and shows that TLR3 is expressed preferentially in intestinal epithelial cells (IEC), CD172a+CD11R1high and CD4+ cells from ileal Peyer's patches. We characterized the inflammatory immune response triggered by TLR3 activation in a clonal porcine intestinal epitheliocyte cell line (PIE cells) and in PIE-immune cell co-cultures, and demonstrated that these systems are valuable tools to study in vitro the immune response triggered by TLR3 on IEC and the interaction between IEC and immune cells. In addition, we selected an immunobiotic lactic acid bacteria strain, Lactobacillus casei MEP221106, able to beneficially regulate the anti-viral immune response triggered by poly(I:C) stimulation in PIE cells. Moreover, we deepened our understanding of the possible mechanisms of immunobiotic action by demonstrating that L. casei MEP221106 modulates the interaction between IEC and immune cells during the generation of a TLR3-mediated immune response.
Collapse
Affiliation(s)
- Shoichi Hosoya
- Food Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|